WO2008000070A1 - Dérivés de l'acide glycyrrhétinique - Google Patents

Dérivés de l'acide glycyrrhétinique Download PDF

Info

Publication number
WO2008000070A1
WO2008000070A1 PCT/CA2007/001127 CA2007001127W WO2008000070A1 WO 2008000070 A1 WO2008000070 A1 WO 2008000070A1 CA 2007001127 W CA2007001127 W CA 2007001127W WO 2008000070 A1 WO2008000070 A1 WO 2008000070A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
cdoda
compound
cells
pparγ
Prior art date
Application number
PCT/CA2007/001127
Other languages
English (en)
Other versions
WO2008000070B1 (fr
Inventor
Stephen H. Safe
Sudhakar Chintharlapalli
Alan Mcalees
Robert Mccrindle
Original Assignee
Wellington Laboratories Inc.
The Texas A & M University System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wellington Laboratories Inc., The Texas A & M University System filed Critical Wellington Laboratories Inc.
Priority to US12/308,798 priority Critical patent/US8389573B2/en
Priority to EP07720042.6A priority patent/EP2032594B1/fr
Priority to JP2009516837A priority patent/JP5610766B2/ja
Priority to CA2653726A priority patent/CA2653726C/fr
Publication of WO2008000070A1 publication Critical patent/WO2008000070A1/fr
Publication of WO2008000070B1 publication Critical patent/WO2008000070B1/fr
Priority to HK09107679.4A priority patent/HK1128477A1/zh
Priority to US13/759,320 priority patent/US20130210926A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J63/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton has been modified by expansion of only one ring by one or two atoms
    • C07J63/008Expansion of ring D by one atom, e.g. D homo steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to novel glycerrhetinic acid derivatives, pharmaceutical compositions comprising said derivatives and their use as therapeutics, in particular as a new class of anticancer drugs that act through multiple pathways.
  • Licorice root extracts have been extensively used for their therapeutic properties which include the potentiation of Cortisol action, inhibition of testosterone biosynthesis, reduction in body fat mass and other endocrine effects (1-4).
  • the activities of these extracts are linked to different classes of phytochemicals particularly the major water soluble constituent glycyrrhizin and its hydrolysis product 78 ⁇ -glycyrrhetinic acid (GA):
  • Glycyrrhizin is a pentacyclic triterpenoid glycoside which is hydrolyzed in the gut to GA and many of the properties of licorice root can be attributed to GA.
  • GA inhibits 11 ⁇ -hydroxysteroid dehydrogenase activity increasing corticosterone levels and this has been linked to apoptosis in murine thymocytes, splenocytes and decreased body fat index in human studies (5-9).
  • GA also directly acts on mitochondria to induce apoptosis through increased mitochondrial swelling, loss of mitochondrial membrane potential and release of cytochrome C (10, 11).
  • GA has also been used as a template to synthesize bioactive drugs.
  • carbenoxolone is the 3-hemisuccinate derivative of GA and this compound has been used for the treatment of gastritis and ulcers (12).
  • carbenoxolone may be due to hydrolysis to GA however carbenoxolone itself induced oxidative stress in liver mitochondria and decreased mitochondrial membrane potential.
  • carboxyl and hydroxyl derivatives of glycyrrhetinic acid inhibit HIV and exhibit anti-inflammatory and immunomodulatory activities (13).
  • GA derivatives containing a reduced carboxylic acid group (CH 2 OH) at C-30 and some additional functional changes exhibited strong antioxidant activity (14).
  • GA is an oleanane derivative and there have been extensive structure- activity studies on the anti-inflammatory activities of oleanolic and ursolic acids derivatives (15-19).
  • Two examples that have been prepared and studied are 2-cyano-3,12-dioxo-oleana-1 , 9(11)-diene-28-oic acid (CDDO) and its methyl ester (CDDO-Me) which contain major structural differences in the E-ring compared to GA:
  • CDDO peroxisome proliferator-activated receptor ⁇
  • PPAR is a member of the nuclear receptor (NR) family of transcription factors (23-27), and the three members of this subfamily serve as regulators of lipid and carbohydrate metabolism and play a critical role in multiple diseases including diabetes, atherosclerosis and cancer.
  • Ligand activation of PPAR ⁇ results in formation of a DNA-bound heterodimer with the retinoic acid X receptor (RXR) and after recruitment of the appropriate nuclear factors, transcriptional activation of target gene expression is observed.
  • RXR retinoic acid X receptor
  • the assembly of a transcriptionally-active PPAR/RXR complexes may be highly variable and dependent on expression of coregulatory proteins, and this may dictate, in part, the tissue-specific and ligand structure-dependent activation of PPAR-mediated gene expression and responses.
  • PPAR ⁇ agonists have been developed for treatment of metabolic diseases, and thiazolidinediones (TZDs) are PPAR ⁇ agonists and are used by millions of patients in the United States for treatment of insulin-resistant Type Il diabetes.
  • PPAR ⁇ is overexpressed in multiple tumor-types (28), and there is evidence that various structural classes of PPAR ⁇ agonists inhibit growth and induce apoptosis in both pancreatic and colon cancer cells and tumors (29-50).
  • it is clear from studies with PPAR ⁇ agonists that their effects in colon, pancreatic and other cancer cell lines and tumors are highly variable and can be mediated through receptor-dependent and -independent pathways. Nevertheless, this characteristic of multiple mechanisms can be advantageous for cancer chemotherapy by targeting several pathways that inhibit tumor growth and metastasis.
  • Sp1 and Sp3 proteins are ubiquitously expressed and have been extensively investigated. For example, Sp1 'A embryos exhibit multiple abnormalities, retarded development and embryolethality on day 11 of gestation (57). Sp3 "A mice also exhibit growth retardation, defects in late tooth development, and the animals die at birth (58, 59). The critical requirement for Sp proteins during embryonic and postnatal development is in contrast to decreased expression in mature tissue/organs which are relatively quiescent.
  • Sp1 the major focus of most studies
  • other Sp proteins such as Sp3 and Sp4 are overexpressed in tumors compared to most other tissues/organs (60-65).
  • a recent study compared the expression of Sp1, Sp3 and Sp4 in prostate and pancreatic tumors in xenograft or orthotopic mouse models, and results illustrated the high expression in LNCaP prostate tumor xenografts vs. normal mouse liver from the same animals (66, 67).
  • Levels of Sp1 , Sp3 and Sp4 expression were barely detectable in liver and other tissues compared to high levels of Sp1 , Sp3 and Sp4 in tumors, and several studies report that Sp proteins are overexpressed in multiple tumors (60-65).
  • Glycyrrhetinic acid has been prepared and shown to inhibit colon, pancreatic and prostate cancer cell growth and to induce peroxisome proliferator-activated receptor ⁇ (PPAR ⁇ ) transactivation as well as to induce specificity (Sp) protein degradation.
  • the present invention therefore includes a novel class of new mechanism-based anticancer drugs that act as PPARy agonists and by decreasing expression of Sp proteins in various tumor cells.
  • one aspect of the present invention includes a compound selected from a compound of formula (I):
  • R 1 is selected from CN, halo, NO 2 , CO 2 R 3 , Ci -6 alkyl, fluoro-substituted Ci- ealkyl, C 2-6 alkenyl, C 2-6 alkynyl, OR 3 , SR 3 , SOR 3 , SO 2 R 3 , NR 3 R 4 , C(O)NR 3 R 4 ,
  • R 2 is selected from OCi -6 alkyl, fluoro-substituted OCi -6 alkyl, NH 2 , NHCi -6 alkyl, N(C 1-6 alkyl)(Ci -6 alkyl), SH and SC 1-6 alkyl;
  • R 3 , R 4 and R 5 are independently selected from H, C 1-6 alkyl, fluoro-substituted
  • the present invention also includes a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable carrier.
  • the present invention also includes a use of a compound of the invention as a medicament or as a diagnositic.
  • a further aspect of the present invention is a use of a compound of the invention to treat a condition or disease that benefits from an upregulation of PPAR ⁇ and/or a downregulation of the expression or activity of one or more specificity (Sp) proteins.
  • the condition or disease that benefits from an upregulation of PPAR ⁇ and/or a downregulation of the expression or activity of one or more specificity Sp proteins is cancer.
  • a method of treating cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof.
  • the invention includes a use of a compound of the invention to treat cancer, as well as a use of a compound of the invention to prepare a medicament to treat cancer.
  • the present invention also includes a method of treating diabetes, comprising administering an effective amount of PPAR ⁇ -upregulating effective amount of a compound of the invention to a subject in need thereof.
  • the invention also includes a use of a PPAR ⁇ -upregulating compound of the invention to treat diabetes as well as a use of a PPAR ⁇ -upregulating compound of the invention to prepare a medicament to treat diabetes.
  • a further aspect of the present invention is a method of treating Huntington's disease comprising administering an Sp protein-downregulating effective amount of a compound of the invention to a subject in need thereof. Also included in the present invention is a use of an Sp protein- downregulating compound of the invention to treat Huntington's disease as well as a use of an Sp protein-downregulating compound of the invention to prepare a medicament to treat Huntington's disease.
  • Figure 1 shows ligand-dependent activation of PPAR ⁇ -GAL4/pGAL4 in SW480 cells.
  • Cells were transfected with PPAR ⁇ -GAL4/pGAL4, treated with different concentrations of the triterpenoids, and luciferase activity was determined as described in the Examples. Results of all transactivation studies in this Figure are presented as means ⁇ SE for at least 3 separate determinations for each treatment group and significant (p ⁇ 0.05) induction compared to solvent (DMSO) control is indicated by an asterisk.
  • Figure 2 shows ligand-dependent activation of PPAR ⁇ -GAL4/pGAL4 in
  • HT-29 cells HT-29 cells.
  • Cells were transfected with PPAR ⁇ -GAL4/pGAI_4, treated with different concentrations of the triterpenoids, and luciferase activity was determined as described in the Examples. Results of all transactivation studies in this Figure are presented as means ⁇ SE for at least 3 separate determinations for each treatment group and significant (p ⁇ 0.05) induction compared to solvent (DMSO) control is indicated by an asterisk.
  • Figure 3 shows inhibition of transactivation in SW480 cells transfected with PPAR ⁇ -GAL4/pGAL4 by PPAR ⁇ antagonists.
  • Cells were transfected with PPAR ⁇ -GAL4/pGAL4, treated with different concentrations of CDODA or CDODA-Me alone or in combination with 10 ⁇ M T007, and luciferase activities were determined as described in Figure 1.
  • Significant (p ⁇ 0.05) inhibition of induced transactivation by T007 is indicated (**).
  • Figure 4 shows inhibition of transactivation in SW480 cells transfected with PPRE 3 -LuC by PPAR ⁇ antagonists.
  • Cells were transfected with PPRE 3 - Luc, treated with different concentrations of CDODA-Me alone or in combination with 10 ⁇ M GW9662 and/or T007, and luciferase activities were determined as described in Figure 1. Significant (p ⁇ 0.05) inhibition of induced transactivation by T007 or GW9662 is indicated (**).
  • FIG. 5 shows ligand-induced PPAR ⁇ -coactivator interactions.
  • SW480 cells were transfected with VP-PPAR ⁇ , coactivator-GAL4/pGAL4, treated with different concentrations of CDODA-Me, and luciferase activity was determined as described in the Examples. Results are expressed as means ⁇ SE for 3 replicate determinations for each treatment group, and significant (p ⁇ 0.05) induction is indicated by an asterisk.
  • Figure 6 shows the effects of CDODA-Me on cell cycle proteins, apoptosis and tumor suppressor genes.
  • SW480 cells were treated with different concentrations of CDODA-Me for 24 hr and various proteins were analyzed by western immunoblot analysis as described in the Examples, ⁇ - actin served as a loading control and results were observed in replicate (2 or more) experiments.
  • Figure 7 shows the effects of CDODA-Me and CDODA on cell cycle proteins, apoptosis and tumor suppressor genes.
  • SW480 cells were treated with different concentrations of CDODA-Me or CDODA for 24 and various proteins were analyzed by western immunoblot analysis as described in the Examples, ⁇ -actin served as a loading control and results were observed in replicate (2 or more) experiments.
  • Figure 8 compares the effects of CDODA-Me and CDDO-Me on cell cycle proteins, apoptosis and tumor suppressor genes.
  • SW480 cells were treated with different concentrations of CDODA-Me or CDDO-Me for 96 hr and various proteins were analyzed by western immunoblot analysis as described in the Examples, ⁇ -actin served as a loading control and results were observed in replicate (2 or more) experiments.
  • Figure 9 shows the effects of CDODA-Me on cell cycle proteins, apoptosis and tumor suppressor genes.
  • ⁇ -actin served as a loading control and results in were observed in replicate (2 or more) experiments.
  • FIG. 10 shows the effects of PPAR ⁇ antagonists on CDODA-Me induced effects on protein expression or apoptosis.
  • SW480 cells were treated for 24 hr with different concentrations of CDODA-Me alone or in combination with 10 ⁇ M T007 and PARP (112 kDa), PARP (85 kDa), CD-1 , p27, NAG-1 and KLF-4 proteins were analyzed by western immunoblots as described in the Examples, ⁇ -actin served as a loading control and results were observed in replicate (2 or more) experiments.
  • Figure 11 shows the effects of PPAR ⁇ antagonists on CDODA-Me induced effects on protein expression or apoptosis.
  • HT-29 cells were treated for 24 hr with different concentrations of CDODA-Me alone or in combination with 10 ⁇ M T007 and KLF-4 protein was analyzed by western immunoblots as described in the Examples, ⁇ -actin served as a loading control and results were observed in replicate (2 or more) experiments.
  • Figure 12 shows the effects of PPAR ⁇ antagonists on CDODA-Me induced effects on protein expression or apoptosis.
  • HT-29 cells were treat for 24 hr with different concentrations of CDODA-Me alone or in combination with 10 ⁇ M GW9662 and PARP (112 kDa), PARP (85 kDa) and NAG-1 proteins were analyzed by western immunoblots as described in the Examples, ⁇ -actin served as a loading control and results were observed in replicate (2 or more) experiments.
  • Figure 13 shows the effects of PPAR ⁇ antagonists on CDODA-Me induced effects on protein expression or apoptosis.
  • HT-29 cells were treated for 96 hr with different concentrations of CDODA-Me alone or in combination with 10 ⁇ M T007 and Cav-1 protein was analyzed by western immunoblots as described in the Examples, ⁇ -actin served as a loading control and results were observed in replicate (2 or more) experiments.
  • Figure 14 shows the inhibition of colon (SW480, top) and pancreatic
  • Figure 15 includes gels showing that ⁇ -CDODA-Me induces Sp protein degradation in Panc28 cells. This response is not reversed by T007 (A) and only a minimal amount of reversal is observed with lactacystin (B).
  • Figure 16 includes gels showing the effects of ⁇ -CDODA-Me cell cycle proteins (A) and NAG-1/ATF-3 and PARP cleavage (B) in Panc28 cells.
  • Figure 17 includes a gel showing that ⁇ -CDODA-Me decreases Sp protein protein expression in RKO cells. These effects are not reversed by T007or proteasome inhibitors.
  • Figure 18 shows the effects of ⁇ -CDODA-Me and related compounds on LNCaP cell survival, activation of PPAR ⁇ , and modulation of cell cycle genes.
  • A Cell survival. LNCaP cells were treated with different concentrations of ⁇ -DODA, ⁇ -DODA-Me or ⁇ -CDODA-Me for 96 hr, and the % cell survival relative to DMSO (solvent control set at 100%) was determined as described in the Examples. Results are expressed as means ⁇ SE for three separate determinations for each treatment group, and significantly (p ⁇ 0.05) decreased survival is indicated ( * ).
  • B ⁇ -CDODA-Me activates PPAR ⁇ .
  • LNCaP cells were treated with ⁇ -CDODA, T007 or their combination, transfected with PPAR ⁇ -GAL4/pGAL4 or PPRE-luc, and luciferase activity determined as described in the Examples. Results are expressed as means ⁇ SE for three replicate determinations for each treatment group, and significant (p ⁇ 0.05) induction by ⁇ -CDODA-Me ( * ) and inhibition after cotreatment with T007 ( ** ) are indicated. Modulation of cell cycle genes by ⁇ -CDODA-Me alone (C) and in combination with T007 (D). Cells were treated as indicated for 24 hr, and whole cell lysates were analyzed by Western blot analysis as described in the Examples.
  • Figure 19 shows that ⁇ -CDODA induces apoptotic pathways and decreases androgen-responsiveness in LNCaP cells.
  • LNCaP cells were treated as indicated for 24 hr, and whole cell lysates were analyzed by Western blot analysis as described in the Examples.
  • ⁇ -CDODA-Me- induced DNA fragmentation (A) was also determined as described. Effects of ⁇ -CDODA-Me alone and in combination with DHT or T007 (C) or MG132 (D), and whole cell lysates were analyzed by Western blot analysis as described in the Examples.
  • Figure 20 shows that ⁇ -CDODA-Me induces proapoptotic proteins and kinases.
  • LNCaP cells were treated with 2.5 ⁇ M ⁇ -CDODA-Me, and whole cell lysates isolated at different times after treatment were analyzed by Western blot analysis as described in the Examples. Effects of kinase inhibitors on proapoptotic responses (C) and quantitation of NAG-1 and ATF-3 expression (D).
  • LNCaP cells were treated with 2.5 ⁇ M ⁇ -CDODA alone or in combination with various kinase inhibitors and after 24 hr, whole cell lysates were analyzed by Western blot analysis.
  • Levels of NAG-1 and ATF-3 proteins (normalized to ⁇ -actin) (D) are means ⁇ SE for three separate determinations for each treatment group and significantly (p ⁇ 0.05) decreased levels after cotreatment with a kinase inhibitor are indicated (* * ).
  • Figure 21 shows that ⁇ -CDODA-Me induction of p21 is MAPK- dependent.
  • A Effects of kinase inhibitors on induction of p21.
  • LNCaP cells were treated with DMSO, 2.5 ⁇ M ⁇ -CDODA-Me alone or in combination with kinase inhibitors for 24 hr, and whole cell lysates were analyzed by Western blot analysis as described in the Examples.
  • B ⁇ -CDODA-Me activates p21 promoter constructs. LNCaP cells were transfected with p21 promoter constructs, treated with DMSO or different concentrations of ⁇ -CDODA-Me, and luciferase activity was determined as described in the Examples. Results are means ⁇ SE for three separate determinations for each treatment group, and significant (p ⁇ 0.05) induction of activity is indicated (*).
  • C Inhibition by PD98059.
  • LNCaP cells were treated with ⁇ -CDODA-Me alone or in combination with T007 or cycloheximide for 12 or 18 hr, and AR mRNA levels were determined by real time PCR as described in the Examples.
  • C ⁇ -CDODA-Me decreases AR promoter activity. LNCaP cells were transfected with AR-luc, treated with DMSO or ⁇ -CDODA-Me, and luciferase activity determined as described in the Examples.
  • Results are means ⁇ SE for three separate experiments for each treatment group and a significant (p ⁇ 0.05) decrease in activity is indicated ( * ).
  • D Time-dependent effects of ⁇ -CDODA-Me on AR, Sp1 and PARP (cleaved). LNCaP cells were treated with DMSO or ⁇ -CDODA-Me for up to 24 hr, and whole cell lysates were analyzed by Western blot analysis as described in the Examples.
  • Figure 23 shows that ⁇ -CDODA-Me decreases PSA expression. Effects of T007 (A) and cycloheximide (B) on ⁇ -CDODA-Me-dependent effects on PSA gene expression. LNCaP cells were treated with ⁇ -CDODA- Me alone or in combination with T007 or cycloheximide for 12 or 18 hr, and PSA mRNA levels were determined by real time PCR as described in the Examples. ⁇ -CDODA-Me decreases PSA promoter (C) and DHT-induced (D) PSA promoter activity.
  • C PSA promoter
  • D DHT-induced
  • LNCaP cells were transfected with PSA-luc, treated with DMSO, ⁇ -CDODA-Me, DHT and ⁇ -CDODA-Me plus DHT (combined), and luciferase activity determined as described in the Examples. Results are means ⁇ SE for three replicate determinations for each treatment group, and significantly (p ⁇ 0.05) decreased basal or DHT-induced luciferase activity by ⁇ -CDODA-Me is indicated (*).
  • the “compounds of the invention” include compounds of Formula I as hereinbefore defined, including all polymorphs and crystal habits thereof, salts, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labeled compounds of Formula I.
  • alkyl when used alone or in combination with other groups or atoms, refers to a saturated straight or branched chain consisting solely of 1 to 6 hydrogen-substituted carbon atoms, suitably 1 to 4 hydrogen-substituted carbon atoms, and includes methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, isobutyl, t-butyl, 2,2-dimethylbutyl, n-pentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, n-hexyl and the like.
  • alkenyl refers to a partially unsaturated straight or branched chain consisting solely of 2 to 6 hydrogen- substituted carbon atoms that contains at least one double bond, and includes vinyl, allyl, 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, 2- methylbut-1-enyl, 2-methylpent-1-enyl, 4-methylpent-1-enyl, 4-methylpent-2- enyl, 2-methylpent-2-enyl, 4-methylpenta-1 ,3-dienyl, hexen-1-yl and the like.
  • alkynyl refers to a partially unsaturated straight or branched chain consisting solely of 2 to 8 hydrogen- substituted carbon atoms that contains at least one triple bond, and includes ethynyl, 1-propynyl, 2-propynyl, 2-methylprop-1-ynyl, 1-butynyl, 2-butynyl, 3- butynyl, 1 ,3-butadiynyl, 3-methylbut-1-ynyl, 4-methylbut-ynyl, 4-methylbut-2- ynyl, 2-methylbut-1-ynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1 ,3- pentadiynyl, 1 ,4-pentadiynyl, 3-methylpent-1-ynyl, 4-methylpent-2-ynyl, 4-methylpent-2-ynyl, 4-methylpent-2-
  • aryl refers to an aromatic mono- or bicyclic group containing from 6 to 14 carbon atoms that may be optionally fused with a fully or partially saturated carbocyclic ring and may optionally be substituted with one or more substituents, suitably one to three substituents, independently selected from fluoro-substituted Ci -4 alkyl, halo, OCi -4 alkyl, fluoro-substituted OC ⁇ alkyl, NO 2 and CN.
  • substituents suitably one to three substituents, independently selected from fluoro-substituted Ci -4 alkyl, halo, OCi -4 alkyl, fluoro-substituted OC ⁇ alkyl, NO 2 and CN.
  • substituents suitably one to three substituents, independently selected from fluoro-substituted Ci -4 alkyl, halo, OCi -4 alkyl, fluoro-substituted OC ⁇ alkyl, NO
  • heteroaryl refers to an aromatic mono- or bicyclic group containing from 5 to 14 carbon atoms, of which one to five is replaced with a heteroatom selected from N, S and O, that may optionally be substituted with one or more substituents, suitably one to three substituents, independently selected from C- ⁇ -4 alkyl, fluoro-substituted halo, OCi -4 alkyl, fluoro-substituted OCi -4 alkyl, NO 2 and CN.
  • aryl groups include thienyl, benzimidazolyl, benzo[6]thienyl, furanyl, benzofuranyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, quinolizinyl, isoquinolyl, quinolyl, and the like.
  • fluoro-substituted means that, in the group being described, one or more, including all, of the hydrogen atoms has been replaced by F.
  • a fluoro-substituted alkyl includes trifluoromethyl, trifluoroethyl, pentafluoroethyl and the like.
  • halogen and “halo” include F, Cl, Br, and I.
  • point of attachment of the designated side chain is described first followed by the adjacent functionality toward the terminal portion.
  • a substituent's point of attachment may also be indicated by a dashed line to indicate the point(s) of attachment, followed by the adjacent functionality and ending with the terminal functionality.
  • pharmaceutically acceptable means compatible with the treatment of animals, in particular, humans.
  • pharmaceutically acceptable salt includes both pharmaceutically acceptable acid addition salts and pharmaceutically acceptable basic addition salts.
  • pharmaceutically acceptable acid addition salt means any non-toxic organic or inorganic salt of any base compound of the disclosure, or any of its intermediates.
  • Basic compounds of the disclosure that may form an acid addition salt include, for example, where the R 1 and/or R 2 is substituted with NH 2 , NHCi-C 6 alkyl or N(Ci-C 6 alkyl)(Cr C ⁇ alkyl).
  • Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acids, as well as metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate.
  • Illustrative organic acids that form suitable salts include mono-, di-, and tricarboxylic acids such as glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic and salicylic acids, as well as sulfonic acids such as p- toluene sulfonic and methanesulfonic acids. Either the mono or di-acid salts can be formed, and such salts may exist in either a hydrated, solvated or substantially anhydrous form.
  • mono-, di-, and tricarboxylic acids such as glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic and salicylic acids, as well as sul
  • the acid addition salts of the compounds of the disclosure are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms.
  • the selection of the appropriate salt will be known to one skilled in the art.
  • Other non-pharmaceutically acceptable acid addition salts e.g. oxalates, may be used, for example, in the isolation of the compounds of the disclosure, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • pharmaceutically acceptable basic salt means any non-toxic organic or inorganic basic addition salt of any acid compound of the invention, or any of its intermediates, which are suitable for or compatible with the treatment of animals, in particular humans.
  • Acidic compounds of the invention that may form a basic addition salt include, for example, those where R 1 is C(O)OH.
  • Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium or barium hydroxide.
  • Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethylamine and picoline or ammonia. The selection of the appropriate salt will be known to a person skilled in the art.
  • non-pharmaceutically acceptable basic addition salts may be used, for example, in the isolation of the compounds of the invention, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • the formation of a desired compound salt is achieved using standard techniques. For example, the neutral compound is treated with a base in a suitable solvent and the formed salt is isolated by filtration, extraction or any other suitable method.
  • cancer refers to a class of diseases or disorders characterized by uncontrolled division of cells and the ability of these cells to invade other tissues, either by direct growth into adjacent tissue through invasion or by implantation into distant sites by metastasis. Metastasis is defined as the stage in which cancer cells are transported through the bloodstream or lymphatic system.
  • cancer that may be treated using the compounds of the invention include those that benefit from an up-regulation of the activity of PPAR ⁇ relative to normal cells and/or that benefit from a downregulation of the expression and/or activity of specificity proteins (Sp), in particular Sp1, Sp3 and/or Sp4.
  • Sp specificity proteins
  • Examples of such cancers include, but are not limited to, prostate cancer, colon cancer, breast cancer, bladder cancer, lung cancer, ovarian cancer, endometrial cancer renal cancer and pancreatic cancer.
  • the cancer is prostate cancer, colon cancer or pancreatic cancer.
  • a "therapeutically effective amount", “effective amount” or a “sufficient amount” of a compound of the present invention is a quantity sufficient to, when administered to the subject, including a mammal, for example a human, effect beneficial or desired results, including clinical results, and, as such, an "effective amount” or synonym thereto depends upon the context in which it is being applied.
  • a mammal for example a human
  • an "effective amount” or synonym thereto depends upon the context in which it is being applied.
  • it is an amount of the compound sufficient to achieve such an upregulation of PPAR ⁇ activity as compared to the response obtained without administration of the compound.
  • down regulating the expression and/or activity of Sp proteins for example, it is an amount of the compound sufficient to achieve such a downregulation as compared to the response obtained without administration of the compound.
  • therapeutically effective amounts of the compounds of the present invention are used to treat, modulate, attenuate, reverse, or affect a disease or conditions that benefits from an upregulation of PPAR ⁇ activity and/or downregulation of the expression and/or activity of Sp proteins, for example, cancer in a subject.
  • An "effective amount” is intended to mean that amount of a compound that is sufficient to treat, prevent or inhibit such diseases or conditions.
  • the amount of a given compound of the present invention that will correspond to such an amount will vary depending upon various factors, such as the given drug or compound, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art.
  • a "therapeutically effective amount" of a compound of the present invention is an amount which prevents, inhibits, suppresses or reduces a disease or conditions that benefits from an upregulation of PPAR ⁇ activity and/or downregulation of the expression and/or activity of Sp proteins, for example, cancer as determined by clinical symptoms or the amount of cancer cells, in a subject as compared to a control.
  • a therapeutically effective amount of a compound of the present invention may be readily determined by one of ordinary skill by routine methods known in the art.
  • a therapeutically effective amount of a compound of the present invention ranges from about 0.1 to about 40 mg/kg body weight, suitably about 1 to about 10 mg/kg body weight, and more suitably, from about 2 to about 5 mg/kg body weight.
  • certain factors may influence the dosage required to effectively treat a subject, or prevent a subject, suffering from a disease or conditions that benefits from an upregulation of PPAR ⁇ activity and/or downregulation of the expression and/or activity of Sp proteins, for example cancer, and these factors include, but are not limited to, the severity of the disease or disorder, previous treatments, the general health and/or age of the subject and other diseases present.
  • a "treatment” or “prevention” regime of a subject with a therapeutically effective amount of the compound of the present invention may consist of a single administration, or alternatively comprise a series of applications.
  • the compound of the present invention may be administered at least once a week.
  • the compound may be administered to the subject from about one time per week to about once daily for a given treatment.
  • the length of the treatment period depends on a variety of factors, such as the severity of the disease, the age of the patient, the concentration and the activity of the compounds of the present invention, or a combination thereof.
  • the effective dosage of the compound used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required.
  • administered contemporaneously means that two substances are administered to a subject such that they are both biologically active in the subject at the same time.
  • the exact details of the administration will depend on the pharmacokinetics of the two substances in the presence of each other, and can include administering one substance within 24 hours of administration of the other, if the pharmacokinetics are suitable. Designs of suitable dosing regimens are routine for one skilled in the art.
  • two substances will be administered substantially simultaneously, i.e. within minutes of each other, or in a single composition that comprises both substances.
  • treatment is an approach for obtaining beneficial or desired results, including clinical results.
  • beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e. not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • “Palliating" a disease or disorder means that the extent and/or undesirable clinical manifestations of a disorder or a disease state are lessened and/or time course of the progression is slowed or lengthened, as compared to not treating the disorder.
  • prevention or “prophylaxis”, or synonym thereto, as used herein refers to a reduction in the risk or probability of a patient becoming afflicted with cancer or manifesting a symptom associated with cancer.
  • To “inhibit” or “suppress” or “reduce” or “downregulate” a function or activity, such Sp protein expression or activity, is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another conditions.
  • a function or activity such as PPAR ⁇ activity
  • PPAR ⁇ activity is to increase the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another conditions.
  • subject or “patient” or synonym thereto, as used herein includes all members of the animal kingdom, especially mammals, including human.
  • the subject or patient is suitably a human.
  • a cell as used herein includes a plurality of cells. Administering a compound to a cell includes in vivo, ex vivo and in vitro treatment.
  • Results show that a 2-cyano derivative of GA, namely methyl 2-cyano-3,11-dioxo-18 ⁇ -olean-1 ,12-dien-30-oate ( ⁇ -CDODA- Me) and the corresponding 18 ⁇ isomer ( ⁇ -CDODA-Me), along with structurally related analogs, both activate PPAR ⁇ and induce Sp protein degradation in colon and pancreatic cancer cells.
  • CDODA-Me and related compounds are a novel class of new mechanism-based anticancer drugs that act as PPAR ⁇ agonists and by decreasing expression of Sp proteins in pancreatic and colon cancer.
  • the present invention includes a compound selected from a compound of Formula (I):
  • R 1 is selected from CN, halo, NO 2 , CO 2 R 3 , Ci -6 alkyl, fluoro-substituted Ci-
  • R 2 is selected from OCi -6 alkyl, fluoro-substituted OC 1-6 alkyl, NH 2 , NHCi -6 alkyl, N(C 1-6 alkyl)(Ci -6 alkyl), SH and SCi -6 alkyl;
  • R 1 is selected from CN, halo, NO 2 , CO 2 H, CO 2 Ci -6 alkyl, Ci -6 alkyl, fluoro-substituted Ci -6 alkyl, C 2- ealkenyl, C 2-6 alkynyl, OCi -6 alkyl, fluoro-substituted OCi -6 alkyl, OH, SH, SCi- 6 alkyl, SOCi -6 alkyl, SO 2 Ci -6 alkyl, NH 2 , NHCi -6 alkyl, N(Ci -6 alkyl)(Ci -6 alkyl), C(O)NH 2 , C(O)NHCi -6 alkyl, C(O)N(Ci -6 alkyl)(Ci -6 alkyl), C(O)C 1-6 alkyl, OC(O)Ci -6 alkyl and NHC(O)Ci -6 alkyl.
  • R 1 is selected from CN, halo, NO 2 , CO 2 H, CO 2 Ci -4 alkyl, Ci -4 alkyl, fluoro-substituted Ci ⁇ alkyl, C 2-4 alkenyl, C 2-4 alkynyl, OCi -4 alkyl, fluoro- substituted OCi- 4 alkyl, OH, SH, SCi -4 alkyl, SOCi -4 alkyl, SO 2 Ci -4 alkyl, NH 2 , C(O)NH 2 , C(O)NHCi -4 alkyl, C(O)N(Ci- 4 alkyl)(Ci- 4 alkyl), C(O)Ci-4alkyl, OC(O)Ci -4 alkyl and NHC(O)Ci- 4 alkyl.
  • R 1 is selected from CN, halo, CO 2 H, CO 2 Ci -4 alkyl, Ci -4 alkyl, fluoro-substituted d ⁇ alkyl, OC 1-4 alkyl, fluoro- substituted OCi -4 alkyl and OH.
  • R 1 is selected from CN, Cl, Br, I, F, CO 2 H, CO 2 CH 3 , CH 3 , CF 3 , OCH 3 , OCF 3 and OH.
  • R 1 is CN, CF 3 or I.
  • R 2 is selected from fluoro-substituted OC 1-4 alkyl, NH 2 , NHC ⁇ alkyl, N(Ci -4 alkyl)(Ci -4 alkyl), SH and SCi -4 alkyl. In further embodiments of the invention R 2 is selected from OCi- 4 alkyl and fluoro-substituted OCi -4 alkyl. In still further embodiments of the invention, R 2 is selected from OCH 2 CH 3 , OCH 3 and OCF 3 . In still further embodiments of the invention, R 2 is OCH 3 .
  • R 3 , R 4 and R 5 are independently selected from H, Ci -4 alkyl, fluoro-substituted C-i ⁇ alkyl and phenyl. In a further embodiment, R 3 , R 4 and R 5 are independently selected from H, methyl and CF 3 .
  • the compounds of Formula I include those having either the ⁇ or ⁇ configuration at carbon 18 or mixtures thereof in any ratio. Accordingly, in an embodiment of the invention, the compound of Formula I is selected from:
  • stereochemistry of the compounds of the invention may be as shown above in any given compound listed herein, such compounds of the invention may also contain certain amounts (e.g. less than 20%, preferably less than 10%, more preferably less than 5%) of compounds of the invention having alternate stereochemistry.
  • the compound of Formula I is selected from: 2-cyano-3,11-dioxo-18 ⁇ -oleana-1 ,12-dien-30-oic acid methyl ester;
  • the compounds of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline.
  • amorphous refers to a state in which the material lacks long range order at the molecular level and, depending upon temperature, may exhibit the physical properties of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid.
  • a change from solid to liquid properties occurs which is characterized by a change of state, typically second order ("glass transition").
  • crystalline refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order ("melting point").
  • the compounds of the invention may also exist in unsolvated and solvated forms.
  • solvate is used herein to describe a molecular complex comprising the compound of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
  • hydrate is employed when said solvent is water.
  • a currently accepted classification system for organic hydrates is one that defines isolated site, channel, or metal-ion coordinated hydrates (see Polymorphism in Pharmaceutical Solids by K. R. Morris, Ed. H. G. Brittain, Marcel Dekker, 1995).
  • Isolated site hydrates are ones in which the water molecules are isolated from direct contact with each other by intervening organic molecules. In channel hydrates, the water molecules lie in lattice channels where they are next to other water molecules.
  • the water molecules are bonded to the metal ion.
  • the complex When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity.
  • the solvent or water When, however, the solvent or water is weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content will be dependent on humidity and drying conditions. In such cases, non- stoichiometry will be the norm.
  • references to compounds of Formula I include references to salts, solvates, prodrugs and multi-component complexes thereof.
  • the compounds of Formula I can be prepared using methods known in the art, for example, 18 ⁇ - and 18 ⁇ -glycyrrhetinic acid and their methyl esters may be converted into the corresponding dienones by reaction with 2- iodoxybenzoic acid as per a reported method (74).
  • the corresponding 1- saturated-2-cyano 18 ⁇ -glycyrrhetinic acid and 1-saturated-2-cyano 18 ⁇ - glycyrrhetinic acid and their methyl esters are known (75) and may be reacted with 2,3-dichloro-5,6-dicyano-1 ,4-benzoquinone (DDQ) to give the corresponding 2-cyano-dienones.
  • DDQ 2,3-dichloro-5,6-dicyano-1 ,4-benzoquinone
  • dienones of 18 ⁇ - and 18 ⁇ - glycyrrhetinic acid and their methyl esters may be iodinated at position 3 by reacting with iodine and pyridine in an ether solvent as described in the Examples herein.
  • the present invention includes radiolabeled forms of the compounds of the invention, for example, compounds of the invention labeled by incorporation within the structure 3 H, 11 C or 14 C or a radioactive halogen such as 125 I and 18 F.
  • a radiolabeled compound of the invention may be prepared using standard methods known in the art.
  • tritium may be incorporated into a compound of the invention using standard techniques, for example by hydrogenation of a suitable precursor to a compound of the invention using tritium gas and a catalyst.
  • a compound of the invention containing radioactive iodo may be prepared from the corresponding trialkyltin (suitably trimethyltin) derivative using standard iodination conditions, such as [ 125 I] sodium iodide in the presence of chloramine-T in a suitable solvent, such as dimethylformamide.
  • standard iodination conditions such as [ 125 I] sodium iodide in the presence of chloramine-T in a suitable solvent, such as dimethylformamide.
  • the trialkyltin compound may be prepared from the corresponding non-radioactive halo, suitably iodo, compound using standard palladium-catalyzed stannylation conditions, for example hexamethylditin in the presence of tetrakis(triphenylphosphine) palladium (0) in an inert solvent, such as dioxane, and at elevated temperatures, suitably 50-100 0 C.
  • a compound of the invention containing a radioactive fluorine may be prepared, for example, by reaction of K[ 18 F]/K222 with a suitable precursor compound, such as a compound of Formula I comprising a suitable leaving group, for example a tosyl group, that may be displaced with the 18 F anion.
  • a suitable precursor compound such as a compound of Formula I comprising a suitable leaving group, for example a tosyl group, that may be displaced with the 18 F anion.
  • the present invention relates to novel compounds of Formula I, accordingly the present invention includes all uses of these compounds including, for example, in therapeutic and diagnostic applications.
  • the present invention accordingly includes the use of a compound of the invention as a medicament or as a diagnositic.
  • certain compounds of the invention are useful for treating any condition or disease that benefits from an upregulation of PPAR ⁇ .
  • the condition or disease that that benefits from an upregulation of PPAR ⁇ is diabetes and cancer.
  • the present invention includes a method of treating cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof.
  • the invention also includes a use of a compound of the invention to treat cancer and a use of a compound of the invention to prepare a medicament to treat cancer.
  • the cancer is selected from prostate cancer and gastrointestinal cancers, for example, colon cancer and pancreatic cancer.
  • the present invention also includes a method of treating cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof. Further the invention includes a use of a compound of the invention to treat cancer, as well as a use of a compound of the invention to prepare a medicament to treat cancer.
  • a method of treating diabetes comprising administering an effective amount of PPAR ⁇ -upregulating effective amount of a compound of the invention to a subject in need thereof.
  • the present invention also includes a use of a PPAR ⁇ -upregulating compound of the invention to treat diabetes as well as a use of a PPAR ⁇ -upregulating compound of the invention to prepare a medicament to treat diabetes.
  • the PPAR ⁇ -upregulating compound is 2-cyano- 3,11-dioxo-18 ⁇ -oleana-1,12-dien-30-oic acid methyl ester.
  • the compounds of the invention are useful for treating any condition or disease that benefits from a downregulation in the expression or activity of Sp proteins.
  • the condition or disease that that benefits from a downregulation in the expression or activity of Sp proteins, in particular Sp1 is Huntington's disease.
  • the benefit provided to the pathology of Huntington's disease by suppressing the expression and/or activity of Sp1 has been reported by Qiu, Z. et al. J. Biol. Chem. 281 :16672, 2006.
  • a method of treating Huntington's disease comprising administering an Sp protein-downregulating effective amount of a compound of the invention to a subject in need thereof.
  • the present invention also includes a use of an Sp protein-downregulating compound of the invention to treat diabetes as well as a use of an Sp protein-downregulating compound of the invention to prepare a medicament to treat diabetes.
  • the Sp protein is Sp 1 , Sp3 and/or Sp4.
  • a person skilled in the art would be able to identify Sp protein-downregulating compounds of the invention by contacting one or more cells with a compound of the invention and assaying for the presence of one or more of the Sp proteins and comparing the levels of Sp proteins in the one or more cells with that of controls. Such methods are known in the art (66, 67) and are described in the Examples hereinbelow.
  • the compounds of the invention are suitably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo.
  • the present invention includes a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable carrier or diluent.
  • compositions containing the compounds of the invention can be prepared by known methods for the preparation of pharmaceutically acceptable compositions which can be administered to subjects, such that an effective quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle.
  • suitable vehicles are described, for example, in Remington's Pharmaceutical Sciences (2003 - 20th edition) and in The United States Pharmacopeia: The National Formulary (USP 24 NF19) published in 1999.
  • the compositions include, albeit not exclusively, solutions of the substances in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH and iso-osmotic with the physiological fluids.
  • the described compounds, salts or solvates thereof may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art.
  • the compositions of the invention may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump or transdermal (topical) administration and the pharmaceutical compositions formulated accordingly.
  • Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.
  • a compound of the invention may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet.
  • the compound of the invention may be incorporated with excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • a compound of the invention may also be administered parenterally.
  • Solutions of a compound of the invention can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. A person skilled in the art would know how to prepare suitable formulations.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersion and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. Ampoules are convenient unit dosages.
  • compositions for nasal administration may conveniently be formulated as aerosols, drops, gels and powders.
  • Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device.
  • the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use.
  • the dosage form comprises an aerosol dispenser, it will contain a propellant which can be a compressed gas such as compressed air or an organic propellant such as fluorochlorohydrocarbon.
  • the aerosol dosage forms can also take the form of a pump-atomizer.
  • compositions suitable for buccal or sublingual administration include tablets, lozenges, and pastilles, wherein the active ingredient is formulated with a carrier such as sugar, acacia, tragacanth, or gelatin and glycerine.
  • Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base such as cocoa butter.
  • compositions for topical administration may include, for example, propylene glycol, isopropyl alcohol, mineral oil and glycerin.
  • Preparations suitable for topical administration include liquid or semi-liquid preparations such as liniments, lotions, applicants, oil-in-water or water-in-oil emulsions such as creams, ointments or pastes; or solutions or suspensions such as drops.
  • the topical preparations may include one or more additional ingredients such as diluents, buffers, flavouring agents, binders, surface active agents, thickeners, lubricants, preservatives, e.g. methyl hydroxybenzoate (including anti-oxidants), emulsifying agents and the like.
  • Sustained or direct release compositions can be formulated, e.g. liposomes or those wherein the active compound is protected with differentially degradable coatings, such as by microencapsulation, multiple coatings, etc. It is also possible to freeze-dry the compounds of the invention and use the lypolizates obtained, for example, for the preparation of products for injection.
  • the compounds of the invention may be administered to a subject alone or in combination with pharmaceutically acceptable carriers, as noted above, and/or with other pharmaceutically active agents for the treatment of psychosis, the proportion of which is determined by the solubility and chemical nature of the compounds, chosen route of administration and standard pharmaceutical practice.
  • the dosage of the compounds of Formula I and/or compositions of the invention can vary depending on many factors such as the pharmacodynamic properties of the compound, the mode of administration, the age, health and weight of the recipient, the nature and extent of the symptoms, the frequency of the treatment and the type of concurrent treatment, if any, and the clearance rate of the compound in the animal to be treated.
  • One of skill in the art can determine the appropriate dosage based on the above factors.
  • the compounds of Formula I may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response. For ex vivo treatment of cells over a short period, for example for 30 minutes to 1 hour or longer, higher doses of compound may be used than for long term in vivo therapy.
  • the compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies, for example other agents or therapies that treat cancer, for example, but not limited to, cytotoxic drugs, kinase inhibitors, antibodies and immunotherapy, selective receptor modulators, non-steroidal anti-inflammatory drugs (NSAIDS) and enzyme modulators
  • Methyl 78y ⁇ glycyrrhetinate was prepared by diazomethylation of 18 ⁇ - glycyrrhetinic acid and a sample (161.6 mg, 0.3333 mmol) reacted with the IBX reagent (373.4 mg, 1.333 mmol, 4 equiv) as described in Example 1 for the parent acid. After a similar work-up, the recovered product (375.7 mg) was triturated with EtOAc, the derived suspension filtered off and washed with more solvent. Evaporation of the combined filtrates gave an off-white solid (256.7 mg) which showed one major band on preparative TLC (MeOH/CH 2 CI 2 ; 1 :19).
  • 2-cyano-3,11-dioxo-78 ⁇ -oleana-1 ,12-dien-30-oic acid was prepared from 78 ⁇ -glycyrrhetinic acid.
  • Methyl 2-cyano-3,11-dioxo-78 ⁇ -oleana-12-en-30-oate was also prepared from methyl 78/?-glycyrrhetinate as previously described (25) and a solution of the ester (246.9 mg, 0.4863 mmol) and DDQ (134.1 mg, 0.5905 mmol) in dry benzene (20 ml_) was heated to reflux for 5 h. The resulting clear solution, on cooling, deposited a fine rust-colored solid, which was filtered off. Evaporation of the filtrate, in vacuo, left a clear orange gum showing one major spot on TLC (EtOAc/hexane; 1 :3).
  • Dimethyl formamide (ca 15 ml_; dried by stirring over CaH 2 overnight under N 2 ) was vacuum-transferred into a dry Schlenk tube containing methyl 3,11-dioxo-2-iodo-18 ⁇ -oleanana-1 ,2-dien-30-oate (Example 5, 216.8 mg, 0.3574 mmol) and cuprous iodide (166.6 mg, 0.8744 mmol). This mixture was allowed to warm up to ambient temperature under vacuum and then N 2 was admitted.
  • reaction solution containing a fine suspension of a rust-coloured solid was allowed to cool and then a saturated aqueous ammonium chloride solution (30 mL) was added.
  • a saturated aqueous ammonium chloride solution (30 mL) was added.
  • the resulting solution was extracted with diethyl ether three times (30, 15, and 15 mL); the rust-coloured solid adhered to the walls of the separating funnel.
  • the combined ether extracts were dried over anhydrous sodium sulphate.
  • SW480 and HT29 Human colon carcinoma cell lines SW480 and HT29 were provided by Dr. Stan Hamilton, M. D. Anderson Cancer Center (Houston, TX); SW-480 and HT-29 cells were maintained in Dulbecco's modified Eagle's medium nutrient mixture with Ham's F-12 (DMEM/Ham's F-12; Sigma-Aldrich, St. Louis, MO) with phenol red supplemented with 0.22% sodium bicarbonate, 0.011 % sodium pyruvate, and 5% fetal bovine serum and 10 ml/l 100x antibiotic antimycotic solution (Sigma-Aldrich). Cells were maintained at 37 0 C in the presence of 5% CO 2 . Antibodies and Reagents
  • Antibodies for poly(ADP-ribose) polymerase, cyclin D1 , p27, p21 , caveolin 1 , KLF4 and Grp78 were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). NAG-1 was from Upstate Biotechnology (Charlottesville, VA). Monoclonal ⁇ -actin antibody was purchased from Sigma-Aldrich. Reporter lysis buffer and luciferase reagent for luciferase studies were supplied by Promega (Madison, Wl).
  • ⁇ -Galactosidase ( ⁇ -Gal) reagent was obtained from Tropix (Bedford, MA), and LipofectAMINE reagent was purchased from Invitrogen (Carlsbad, CA).
  • Western Lightning chemiluminescence reagent was from PerkinElmer Life and Analytical Sciences (Boston, MA).
  • the PPARy antagonists 2-chloro-5-nitro-N- phenylbenzamide (GW9662) and N-(4'-aminopyridyl)-2-chloro-5- nitrobenzamide (T007) were synthesized using the method described in Chem. Biol. 1997, 4(12):909-918, and their identities and purity (>98%) were confirmed by gas chromatography-mass spectrometry. Plasmids
  • Gal4 reporter containing 5x Gal4 response elements was kindly provided by Dr. Marty Mayo (University of North Carolina, Chapel Hill, NC).
  • Gal4DBD-PPAR ⁇ construct (gPPAR ⁇ ) was a gift of Dr. Jennifer L. Oberfield (GlaxoSmithKline Research and Development, Research Triangle Park, NC).
  • PPRE 3 -IuC construct contains three tandem PPREs with a minimal TATA sequence in pGL2.
  • Colon Cancer cell lines SW480 and HT29 (1 x 10 5 cells/well) were plated in 12-well plates in DMEM/Ham's F-12 media supplemented with 2.5% charcoal-stripped FBS. After 16 h, various amounts of DNA [i.e., Gat ⁇ Luc (0.4 ⁇ g), ⁇ -Gal (0.04 ⁇ g), Gal4PPAR and PPRE 3 -LuC (0.04 ⁇ g)] were transfected using LipofectAMINETM reagent (Invitrogen) following the manufacturer's protocol. Five hours after transfection, the transfection mix was replaced with complete media containing either vehicle (DMSO) or the indicated ligand for 20 to 22 h.
  • DMSO vehicle
  • Cells were then lysed with 100 ml of 1x reporter lysis buffer, and 30 ⁇ l of cell extract was used for luciferase and ⁇ -galactosidase assays. Lumicount was used to quantitate luciferase and ⁇ -galactosidase activities, and the luciferase activities were normalized to ⁇ -galactosidase activity.
  • SW480 and HT 29 Cells (2 x 10 4 ) were plated in 12-well plates, and media were replaced the next day with DMEM/Ham's F-12 media containing 2.5% charcoal-stripped FBS and either vehicle (DMSO) or the indicated ligand and dissolved in DMSO. Fresh media and compounds were added every 48 h. Cells were counted at the indicated times using a Coulter Z1 cell counter. Each experiment was done in triplicate, and results are expressed as means ⁇ S. E. for each determination Western Blot Analysis SW-480 and HT-29 (3 x 10 5 ) cells were seeded in six-well plates in
  • Whole-cell lysates were obtained using high-salt buffer [50 mM HEPES, 500 mM NaCI, 1.5 mM MgCI 2 , 1 mM EGTA, 10% glycerol, and 1% Triton X-100, pH 7.5, and 5 ⁇ l/ml Protease Inhibitor Cocktail (Sigma-Aldrich)].
  • Protein samples were incubated at 100 0 C for 2 min, separated on 10% SDS-PAGE at 120 V for 3 to 4 h in 1 x running buffer (25 mM Tris-base, 192 mM glycine, and 0.1% SDS, pH 8.3), and transferred to polyvinylidene difluoride membrane (PVDF; Bio-Rad, Hercules, CA) at 0.1 V for 16 h at 4 0 C in 1x transfer buffer (48 mM Tris-HCI, 39 mM glycine, and 0.025% SDS).
  • PVDF polyvinylidene difluoride membrane
  • the PVDF membrane was blocked in 5% TBST-Blotto (10 mM Tris-HCI, 150 mM NaCI, pH 8.0, 0.05% Triton X-100, and 5% nonfat dry milk) with gentle shaking for 30 min and was incubated in fresh 5% TBST-Blotto with 1 :1000 (for caveolin-1 , p27, p21 , cyclin D1 , Grp78), 1 :500 (for KLF4, NAG-1), 1 :250 (for PARP), and 1 :5000 (for ⁇ -actin) primary antibody overnight with gentle shaking at 4 0 C.
  • 5% TBST-Blotto 10 mM Tris-HCI, 150 mM NaCI, pH 8.0, 0.05% Triton X-100, and 5% nonfat dry milk
  • the PVDF membrane was incubated with secondary antibody (1 :5000) in 5% TBST-Blotto for 90 min.
  • the membrane was washed with TBST for 10 min, incubated with 10 ml of chemiluminescence substrate (PerkinElmer) for 1.0 min, and exposed to Kodak X-OMAT AR autoradiography film (Eastman Kodak, Rochester, NY).
  • ⁇ -DODA ⁇ -DODA
  • ⁇ -CDODA Example 3(a)
  • their corresponding methyl ester derivatives Examples 2(a) and 4(a)
  • the IC 50 values for ⁇ -DODA and ⁇ -DODA-Me were 25 and 10 ⁇ M respectively in SW480 cells and in HT-29 cells, IC 5 O values were similar (20- 30 and 5-10 ⁇ M) respectively).
  • the 2-cyano substituted analogs were more potent inhibitors of SW480 cell proliferation with IC 5 O values of 2.5-5.0 and 0.2 and 0.5 ⁇ M for ⁇ -CDODA and ⁇ -CDODA-Me respectively.
  • IC 50 values for ⁇ -CDODA and ⁇ -CDODA-Me in HT-29 cells were 1.0 and 0.2 to 0.5 ⁇ M respectively indicating that this cell line was more sensitive than SW480 cells to the growth inhibitory effects of ⁇ -CDODA.
  • Previous studies showed that both CDDO and CDDO-Me induced luciferase activity in SW480 cells transfected with GAL4-PPAR ⁇ /GAL4-Luc (22) and the results in Figure 1 summarize the activation of PPAR ⁇ by the GA derivatives of the present invention.
  • ⁇ -CDODA-Me (1-5 ⁇ M) significantly activated PPAR ⁇ with a maximal 18-Fold induction of luciferase activity, whereas 20-30 ⁇ M ⁇ -CDODA induced a ⁇ 4.5 fold increase in activity and up to 30 ⁇ M ⁇ -DODA and ⁇ - DODA-Me did not enhance transactivation.
  • Caveolin-1 acts as a tumor suppressor gene in colon cancer cells and inhibits cell/tumor (in vivo) growth (76, 77). Caveolin-1 is only induced in colon cancer cells after prolonged treatment with PPAR ⁇ agonists and the results in Figure 8 show that although both C D DO/CD DO-Me induce caveolin- 1 protein after treatment for 3 days, ⁇ -CDODA-Me did not affect expression of caveolin-1 in SW480 cells. This was observed over several replicate experiments and clearly distinguished ⁇ -CDODA-Me from CDDO/CDDO-Me in SW480 cells.
  • KLF4 is induced by ⁇ -CDODA-Me in both SW480 and HT-29 cells and a recent study reported that the PPAR ⁇ agonist 15-deoxy- ⁇ 12, 14- Prostaglandin J2 (PGJ2) also induced KLF4 in HT-29.
  • PGJ2 14- Prostaglandin J2
  • MAPK mitogen- activated protein kinase
  • T007 does not affect ⁇ -CDODA-Me induced down regulation of cyclin D1 , p27 or PARP cleavage in SW480 cells ( Figure 10) and the PPARy-independent induction of apoptosis in SW480 cells has previously been reported for CDDO-Me in the same cell line (22).
  • Result in Figure 12 show that induction of NAG-1 and PARP cleavage in HT- 29 cells was not affected after cotreatment with T007 whereas the induction of caveolin-1 by ⁇ -CDODA-Me was inhibited after cotreatment with T007 ( Figure 13).
  • PPARY and other members of the nuclear receptor superfamily are characterized by their modular structure which contains several regions and domains that are required for critical receptor-protein and receptor-DNA interactions (78-79).
  • Nuclear receptors typically contain N- and C- terminal activation functions (AF1 and AF2 respectively), a DNA binding domain and a flexible hinge region.
  • AF1 and AF2 N- and C- terminal activation functions
  • the addition of receptor ligand usually results in formation of a transcriptionally active nuclear receptor complex which binds cognate response elements in promoter regions of target genes and activates transcription.
  • receptor-mediated transactivation is dependent on several factors including cell context-specific expression of coregulatory proteins (eg. coactivators), gene promoter accessibility and ligand structure (80).
  • the complex pharmacology of receptor ligands is due, in part to their structure-dependent conformational changes in the bound receptor complex which may differentially interact with coregulatory factors and exhibit tissue- specific agonist and/or antagonist activity (80, 81).
  • SRMs selective receptor modulators
  • PPAR ⁇ agonists are also SRMs and induce tissue-specific receptor-dependent and independent responses.
  • NAG-1 in HCT116 colon cancer cells was PPAR ⁇ -dependent whereas both troglitazone and PPAR ⁇ -active 1 ,1 -bis (3'-indolyl)-1-(p-substituted phenyl) methanes (C-DIMs) also enhanced NAG-1 expression through receptor-independent pathways in the same cell line (22, 82, 83).
  • ⁇ -CDODA and ⁇ -CDODA-Me the carboxyl substituent is at C- 20 instead of C-17 for CDDO/CDDO-Me, the stereochemistry of the E-D ring fusion at C-18 and the ⁇ , ⁇ -unsaturated ketone moieties in the C-ring are also different in the GA derivatives compared to CDDO.
  • CDDO-Me was active at lower doses than ⁇ -CDODA-Me in both the growth inhibition and transactivation assays in SW480 cells.
  • ⁇ -CDODA was less potent than either ⁇ -CDODA-Me or CDDO in these same assays and therefore was primarily used for further studies.
  • ⁇ -CDODA-Me decreased SW480 and HT-29 cell growth and induced PPAR ⁇ -independent PARP cleavage in both cell lines.
  • ⁇ -CDODA-Me was less potent than CDDO-Me in SW480 cells (22) nevertheless, the newly synthesized GA derivative was a potent anticancer agent in colon cancer cells with effects on cell survival and apoptosis in the higher nM and lower ⁇ M range.
  • ⁇ -CDODA-Me induced the tumor suppressor gene KLF4 in both SW480 and HT-29 cells and this response was PPAR ⁇ -dependent and inhibited by T007.
  • ⁇ -CDODA-Me and PGJ2 which also induced KLF4 in HT29 cells through a receptor- independent pathway (89).
  • Differences between ⁇ -CDODA-Me and CDDO- Me in were observed SW480 cells.
  • CDDO-Me but not ⁇ -CDODA-Me induces caveolin-1 in SW480 cells (22) ( Figure 8) whereas both compounds induce caveolin-1 in HT-29 cells (22) ( Figure 13) and induction of caveolin-1 was inhibited by GW9662.
  • CDODA-Me represents a new class of selective PPAR ⁇ modulators that induces both PPAR ⁇ -dependent and independent responses in colon cancer cells.
  • a previous report showed that KLF4 expression in colon cancer cells was regulated by over expression of the adenomatous polyposis coli gene and by the tumor suppressor homeodomain protein CDX2 (42).
  • APC also enhanced CDX2 expression suggesting an APC-CDX2- KLF4 sequence for activation of KLF4.
  • KLF4 expression is also enhanced by ⁇ -CDODA-Me through a PPAR ⁇ -dependent pathway.
  • Example 8 Effects of compounds of the invention on pancreatic cell lines
  • FIG. 14 illustrates the growth inhibitory effects of ⁇ - and ⁇ -CDODA-Me in Panc28 pancreatic cancer cells, alongside SW580colon cancer cells for reference.
  • the IC 50 values for ⁇ - and ⁇ -CDODA-Me were 0.5 and 0.2-0.5 ⁇ M, respectively, in SW480 cells and 0.5-1.0 and 1-2.5 ⁇ M in Panc28 pancreatic cancer cells.
  • Example 9 Effects of compounds of the invention on Sp protein degradation Sp proteins such as Sp1 , Sp3 and Sp4 are highly expressed in cancer cells, and Sp1 is overexpressed in multiple tumors compared to non-tumor tissue.
  • RNA interference experiments using small interfering RNAs for Sp1 (iSp1), Sp3 (iSp3) and Sp4 (iSp4) that these proteins are required for cell cycle progression, angiogenesis and survival (72, 67).
  • COX-2 inhibitors celecoxib, tolfenamic acid and structurally related NSAIDs
  • betulinic acid activates Sp protein degradation in prostate cancer cells and tumors and this is accompanied by decreased Sp- dependent expression of survivin (antiapoptotic), VEGF (angiogenic) and cell cycle genes.
  • VEGF receptor 1 VEGFR1
  • VEGFR2 VEGF receptor 1
  • ⁇ -CDODA-Me part of the underlying mechanism of action of ⁇ -CDODA-Me is also due to Sp protein degradation.
  • Results in Figure 15 show that after treatment of Panc28 cells with ⁇ -CDODA-Me, there was a concentration-dependent decrease in Sp1 , Sp3 and Sp4 protein expression in Panc28 cells, and this was accompanied by a parallel decrease in VEGF expression and induction of caspase-dependent apoptosis (PARP cleavage) ( Figure 16).
  • Example 9 Effects of the compounds of the invention on prostate cancer cell lines
  • Materials and Methods Cell lines LNCaP human prostate carcinoma cells were obtained from
  • Fetal bovine serum was obtained from JRH Biosciences, Lenexa, KS.
  • LNCaP cells were maintained in RPMI 1640 (Sigma Chemical, St. Louis, MO) supplemented with 0.22% sodium bicarbonate, 0.011% sodium pyruvate, 0.45% glucose, 0.24% HEPES, 10% FBS, and 10 mL/L of 100X antibiotic/antimycotic solution (Sigma). Cells were maintained at 37°C in the presence of 5% CO 2 .
  • Antibodies and Reagents Antibodies for poly(ADP-ribose) polymerase, cyclin D1 , p27, FKBP51, AR, ATF3, Akt and caveolin-1 were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). PSA was obtained from Dako Denmark A/S (Glostrup, Denmark); NAG-1 was purchased from Upstate Biotechnology (Charlottesville, VA); and EGR-1, pAKT, pERK, ERK, pJNK, JNK were obtained from Cell Signaling Technology Inc. (Danvers, MA). Monoclonal ⁇ -actin antibody and dihydrotesterone were purchased from Sigma-Aldrich.
  • Reporter lysis buffer and luciferase reagent for luciferase studies were purchased from Promega (Madison, Wl).
  • ⁇ - Galactosidase ( ⁇ -Gal) reagent was obtained from Tropix (Bedford, MA), and lipofectamine reagents were supplied by Invitrogen (Carlsbad, CA).
  • Western Lightning chemiluminescence reagents were from Perkin-Elmer Life Sciences (Boston, MA).
  • the PPAR ⁇ antagonist N-(4'-aminopyridyl)-2-chloro-5- nitrobenzamide (T007) was prepared in this laboratory and the synthesis of the GA derivatives has previously described (90).
  • LNCaP prostate cancer cells (2 x 10 4 per well) were added to 12-well plates and allowed to attach for 24 hr. The medium was then changed to DMEM/Ham's F- 12 media containing 2.5% charcoal-stripped FBS, and either vehicle (DMSO) or the indicated C-DIMs were added. Fresh medium and indicated compounds were added every 48 hr, and cells were then trypsinized and counted after 2, 4, and 6 days using a Coulter Z1 cell counter (Beckman Coulter, Fullerton, CA). Each experiment was done in triplicate, and results are expressed as means ⁇ S. E. for each set of three experiments. The DNA fragmentation assay was performed using a BioVision Apoptotic DNA ladder extraction kit (BioVision, Mountain View, CA) according to the manufacturer's protocol.
  • the Gal4 reporter construct containing 5X Gal4 response elements was kindly provided by Dr. Marty Mayo (University of North Carolina, Chapel Hill, NC).
  • the Gal4DBD-PPAR ⁇ construct was a gift of Dr. Jennifer L. Oberfield (Glaxo Wellcome Research and Development, Research Triangle Park, NC).
  • the PPRE-luc construct contains three tandem PPREs with a minimal TATA sequence linked to the luciferase gene in pGL2.
  • the AR-luc construct containing the -5400 to +580 region of the androgen receptor promoter was provided by Dr. Donald J.
  • Transfection studies were performed using 0.4 ⁇ g of Gal4Luc, 0.04 ⁇ g of ⁇ -galactosidase, 0.04 ⁇ g of Gal4DBD-PPAR ⁇ , 0.4 ⁇ g of AR-luc, and 0.3 ⁇ g of PSA-luc.
  • the transfection mix was replaced with complete media containing either vehicle (DMSO) or the indicated ligand for 20 to 22 hr.
  • Cells were then lysed with 100 ⁇ l of 1 x reporter lysis buffer, and 30 ⁇ l of cell extract was used for luciferase and ⁇ - galactosidase assays.
  • a Lumicount luminometer (PerkinElmer Life and Analytical Sciences) was used to quantify luciferase and ⁇ -galactosidase activities, and the luciferase activities were normalized to ⁇ -galactosidase activity.
  • PCR was carried out in triplicate in a 25- ⁇ l volume using SYBR Green Master mix (Applied Biosystems) for 15 min at 95°C for initial denaturing, followed by 40 cycles of 95°C for 30 s and 60 0 C for 1 min in the ABI Prism 7700 sequence detection system (Applied Biosystems).
  • the ABI Dissociation Curves software was used after a brief thermal protocol (95°C 15 s and 6O 0 C 20 s, followed by a slow ramp to 95°C) to control for multiple species in each PCR amplification.
  • the comparative CT method was used for relative quantitation of samples. Values for each gene were normalized to expression levels of TATA-binding protein. Primers were purchased from Integrated DNA Technologies (Coralville, IA).
  • sequences of the primers used for reverse transcription-PCR were as follows: AR forward, 5'-GTA CCC TGG CGG CAT GGT-3' [SEQ ID NO: 1] and AR reverse, 5'-CCC ATT TCG CTT TTG ACA CA-3 1 [SEQ ID NO: 2]; PSA forward, 5'-GCA TTG AAC CAG AGG AGT TCT TG-3 1 [SEQ ID NO: 3] and PSA reverse, 5'-TTG CGC ACA CAC GTC ATT G- 3' [SEQ ID NO: 4]; and TATA-binding protein forward, 5'-TGC ACA GGA GCC AAG AGT GAA-3' [SEQ ID NO: 5] and reverse, 5'-CAC ATC ACA GCT CCC CAC CA-3 1 [SEQ ID NO: 6].
  • PVDF polyvinylidene difluoride membrane
  • the membranes were blocked for 30 min with 5% TBST-Blotto (10 mM Tris-HCI, 150 mM NaCI (pH 8.0), 0.05% Triton X-100 and 5% non-fat dry milk) and incubated in fresh 5% TBST-Blotto with primary antibody overnight with gentle shaking at 4 0 C. After washing with TBST for 10 min, the PVDF membrane was incubated with secondary antibody (1 :5000) in 5% TBST-Blotto for 2-3 hr.
  • 5% TBST-Blotto 10 mM Tris-HCI, 150 mM NaCI (pH 8.0), 0.05% Triton X-100 and 5% non-fat dry milk
  • Statistical Analysis Statistical differences between different groups were determined by ANOVA and Scheffe's test for significance. The data are presented as mean ⁇ S. E. for at least three separate determinations for each treatment group.
  • ⁇ -CDODA-Me significantly induced luciferase activity ( Figure 18B) and in cells cotreated with ⁇ -CDODA-Me plus 10 ⁇ M T007 (a PPAR ⁇ antagonist), there was significant inhibition of induced transactivation.
  • ⁇ -DODA-Me and ⁇ -l-DODA- Me did not activate PPAR ⁇ .
  • PPAR ⁇ agonists typically modulate expression of one or more of the cell cycle proteins p27, p21 and cyclin D1
  • Figure 18C illustrates the effects of 1-5 ⁇ M ⁇ -CDODA-Me on expression of these proteins in LNCaP cells.
  • NAG-1 and ATF-3 are proapoptotic proteins induced by PPAR ⁇ agonists and results in Figure 19 show that 1-5 ⁇ M ⁇ -CDODA-Me induced NAG-1 and ATF-3 which are often co-induced and this was accompanied by caspase-dependent PARP cleavage, DNA fragmentation, and decreased bcl2 expression in LNCaP cells.
  • ⁇ -CDODA-Me plus T007 Figure 19B
  • the induced responses were not inhibited by the PPAR ⁇ antagonist indicating that induction of these proapototic responses was receptor-independent.
  • FIG. 19C summarizes the effects of ⁇ -CDODA-Me on AR expression in the presence or absence of 10 nM DHT and also on the expression of FKBP51 and PSA, two androgen-responsive genes in LNCaP cells.
  • DHT increases expression of AR due to stabilization of the receptor and also induces both androgen- responsive FKBP51 and PSA genes and, in cells treated with 1-5 ⁇ M ⁇ - CDODA-Me, there was a concentration-dependent decrease in AR, PSA and FKBP51 expression in the presence or absence of DHT.
  • Figure 2OA summarizes the time-dependent induction of EGR-1 , ATF-3 and NAG-1 by 2.5 ⁇ M ⁇ -CDODA-Me and the induction responses followed a similar time course, whereas EGR-1 dependent induction of NAG-1 in colon cancer cells is associated with the increased expression of EGR-1 prior to induction of NAG-1 (94, 100).
  • Previous studies show that NAG-1 induction is kinase-dependent (94, 100), and results in Figure 2OB show that 2.5 ⁇ M ⁇ -CDODA-Me induces activation of the JNK (P-JNK), PI3K (p-Akt) and MAPK (p-Erk) pathways.
  • Results in Figure 21B show that the 1-5 ⁇ M ⁇ -CDODA-Me also induces luciferase activity in LNCaP cells transfected with constructs containing -2325 to +8 [p21-Luc (Fl)], -124 to +8 [p21-Luc (-124)], -101 to +8 [p21-Luc (-101)], and -60 to +8 [p21-Luc (-60)] p21 promoter inserts.
  • the latter 3 constructs contain the 6 proximal GC rich site (Vl - 1) and the results of the transfection studies suggest that these GC- rich sites are necessary for ⁇ -CDODA-Me-induced transactivation.
  • Deletion analysis of the p21 promoter indicates that loss of inducibility [i.e. p21-luc(60)] is associated with loss of GC-rich sites IV and III which are essential for MAPK-dependent activation of p21 by ⁇ -CDODA-Me.
  • the role of MAPK in activation of the p21 promoter was confirmed in LNCaP cells transfected with p21-luc(101); ⁇ -CDODA-Me induced luciferase activity and cotreatment with the MAPK inhibitor PD98059 inhibited this response (Figure 21C).
  • PSA protein expression is also decreased in LNCaP cells treated with ⁇ -CDODA-Me (Figure 19C) and similar effects were observed for PSA mRNA levels after treatment for 12 or 18 hr, and these responses were not inhibited after cotreatment with the PPAR ⁇ antagonist T007 ( Figure 23A).
  • ⁇ - CDODA-Me-induced downregulation of PSA mRNA levels after treatment for 12 or 18 hr was significantly inhibited after cotreatment with cycloheximide (Figure 23B).
  • ⁇ -CDODA-Me inhibited transactivation in LNCaP cells transfected with the PSA-Luc construct (contains 5.85 kb of the PSA promoter insert) (Figure 23C) and similar results were obtained for DHT- induced luciferase activity ( Figure 23D).
  • ⁇ -CDODA-Me inhibits PSA expression through induction of inhibitory frans-acting factors. Discussion
  • CDODA-Me in LNCaP cells and the role of PPAR ⁇ in mediating these responses was investigated.
  • ⁇ -CDODA-Me was a more potent inhibitor of LNCaP cell growth than analogs ( ⁇ -DODA and ⁇ -DODA-Me) that did not contain a 2-substituent
  • ⁇ -CDODA-Me also activated PPAR ⁇ - dependent transactivation in transient transfection studies in LNCaP cells, and compounds without the 2-substituent were inactive as reported above for these analogs in colon cancer cells.
  • ⁇ -CDODA-Me induced p27 expression and downregulated levels of cyclin D1 protein.
  • ⁇ -CDODA-Me induced p21 protein in LNCaP cells and this response was not inhibited after cotreatment with PPAR ⁇ antagonist T007.
  • ⁇ -CDODA-Me induction of p21 in LNCaP cells was due to activation of MAPK signaling which was required for induction of p21 protein and activation of the p21 promoter. This is a novel pathway for induction of p21 in LNCaP cells; however, previous studies in other cell lines also demonstrated MAPK-dependent induction of p21 expression (101 , 103, 104).
  • NAG-1 and ATF3 are growth inhibitory and proapoptotic proteins (48, 49), and previous studies with PPAR ⁇ agonists report both receptor- dependent and -independent induction of NAG-1 (22, 94, 98, 99). Induction of NAG-1 and ATF3 by ⁇ -CDODA-Me in LNCaP cells was also PPAR ⁇ - independent.
  • the compounds of the present invention decrease expression of Sp1 , Sp3 and Sp3 in colon and pancreatic cancer cells. This correlates with the cytotoxicity, antiangiogenic and proapoptotic effects of these agents. Moreover, ⁇ -CDODA-Me is also a PPAR ⁇ agonist and at least in HCT- 15 colon cancer cells, there is evidence that activation of this pathway is important for the observed anticancer activity. Therefore, the compounds of the present invention appear to inhibit growth of colon, pancreatic and prostate tumor growth through activation of PPAR ⁇ and/or degradation of Sp1 , Sp3 and Sp4 in tumors. The anticancer activity and the tumor and tissue/cell specificity of Sp protein knockdown of the compounds of the invention can be further demonstrated in animal models.
  • the experimental design utilizes the athymic nude mouse xenograft and orthotopic models for prostate, colon and pancreatic cancer, the Min mouse model for colon cancer and the TRAMP model for prostate cancer.
  • SW480, RKO, Panel , PC3 and LNCaP cancer cells (xenograft) and L3.6pl pancreatic cancer cells (xenograft and orthotopic) are used in athymic nude mice and the antitumorigenic effects of the compounds of the invention are investigated.
  • the Min mouse model for colon cancer and the TRAMP model for prostate cancer are used to assay the effects of the compounds of the invention on tumor formation and growth and the determination of selected proapoptotic/antiangiogenic markers are compared to those investigated in the xenografts/orthotopic experiments.
  • mice Male athymic nude mice are obtained from commercial sources and their use approved by the Institutional Animal Care and Use Committee. The mice are housed under specific conditions and in facilities approved by the American Association for Accreditation of Laboratory Animal Care at LARR facilities in College Station, TX, and the corresponding facilities at IBT in Houston, TX. Ten animals are used for each treatment group. SW480, RKO and Panel cells are used in the xenograft study and L3.6pl pancreatic cancer cells are used in the orthotopic model as previously described (66, 67). Cells are harvested by exposure to trypsin and resuspended in serum-free Hanks' balanced salt solution (HBSS).
  • HBSS Hanks' balanced salt solution
  • Viability is assessed by trypan blue exclusion, and only single-cell suspensions exhibiting greater than 95% viability are used.
  • tumor cells (1 x 10 6 cells) suspended in a volume of 200 ⁇ L are implanted subcutaneously in the flank of nude male animals using a 27-gauge needle. Tumors are allowed to grow unperturbed for 10-14 days and when palpable tumors (200 mm 3 ) first appear, mice are randomly assigned to treatment or control groups. Mice are treated (10 per treatment group) with placebo or a compound of the invention (2, 10, or 20 mg/kg/d) (in corn oil) administered every second day for 4 to 6 weeks (depending on appearance and size of control tumors). The doses of the compounds of the invention are estimated from relative potency data.
  • a similar does regiment is used for the orthotopic model for pancreatic cancer using L3.6pl cells as previously described (86). Seven days after implantation of tumor cells into the pancreas of each mouse, 5 mice are sacrificed to confirm the presence of tumor lesions. Compounds are administered three times weekly (i.p. injection). Mice are sacrificed on day 35 and body weights, determined. Primary tumors in the pancreas are excised, measured and weighed. For IHC and H&E staining procedures, one part of the tumor tissue is fixed in formalin and embedded in paraffin, and another part is embedded in OCT compound, rapidly frozen in liquid nitrogen, and stored at -7O 0 C.
  • lmmunohistochemical and Western blot analysis Tumor sections from compound- and corn oil-treated animals are also prepared for in situ hybridization and immunohistochemical analysis of proteins and in situ hybridization (for mRNAs), including proapoptotic (survivin, PARP and caspase 3 cleavage) and angiogenic (VEGF, VEGFR1 and VEGFR2) genes/proteins or responses.
  • immunostaining for Sp1 , Sp3 and Sp4 is done and many of these responses have been determined in previous studies (66, 67, 69-72).
  • At least ten animals are used in each treatment group, and after the last dose, blood is taken and the following parameters determined in a diagnostic laboratory: AST, ALP, LDH, BUN, creatinine, triglycerides, glucose, and total protein.
  • the suppression of lipid levels is a measure of the hyperlipidemic effects which are typically observed for PPAR ⁇ agonists.
  • the intestines are dissected into proximal, middle and distal sections and examined for polyp formation by a veterinary pathologist.
  • expression of Sp proteins and Sp-dependent genes are determined in intestinal tissues/polyps to determine the role of Sp protein degradation in mediating the anticancer responses observed in the Min mouse model.
  • (i) Animal treatment Male athymic nude mice are obtained from commercial sources and their use approved by the Institutional Animal Care and Use Committee. Ten animals are used for each treatment group and based on consultation with biostatisticians, this number is sufficient for determining statistical significance between treatment groups (41 , 42).
  • At least one AR- positive (LNCaP/22Rv1) and AR-negative (PC3/DU145) prostate cancer cell line is used in the xenograft study. Cells are harvested by exposure to trypsin and resuspended in serum-free Hanks' balanced salt solution (HBSS). Viability is assessed by trypan blue exclusion, and only single-cell suspensions exhibiting greater than 95% viability are used.
  • HBSS Hanks' balanced salt solution
  • Tumor cells (1 x 10 6 cells) suspended in a volume of 200 ⁇ l_ are implanted subcutaneously in the flank of nude male animals using a 27-gauge needle. Tumors are allowed to grow unperturbed for 10-14 days and when palpable tumors (200 mm 3 ) first appear, mice are randomly assigned to treatment or control groups. Mice are treated (10 per treatment group) with placebo or a compound of the invention (2, 10, or 20 mg/kg/d) (in corn oil) administered every second day for 4 to 6 weeks (depending on appearance and size of control tumors).
  • Tumor sections from compound- and corn oil-treated animals are also prepared for in situ hybridization and immunohistochemical analysis of proteins and in situ hybridization (for mRNAs), including proapoptotic (survivin, PARP and caspase 3 cleavage) and angiogenic (VEGF, VEGFR1 and VEGFR2) genes/proteins or responses.
  • proapoptotic survivin, PARP and caspase 3 cleavage
  • VEGF angiogenic gene/proteins or responses.
  • immunostaining for Sp1 , Sp3 and Sp4 is done as previously described (41 , 42).
  • Western blot analysis of Sp proteins, proapoptotic and antiangiogenic responses is determined on whole cell lysates from compound- and corn oil (vehicle)- treated tumors.
  • TRAMP mouse model Animal treatment: The TRAMP mouse model is ideal for testing the antitumorigenic activity of the compounds of the invention. Compounds in corn oil are administered every second day from the age of 16 weeks until they are 28 weeks of age when TRAMP mice exhibit approximately 100% primary prostate tumors and metastases.
  • Prostate tumor formation and metastasis Treated and control (corn oil) TRAMP mice are sacrificed at 28 weeks of age and prostate tumor weights, and other organ and whole body weights are recorded; lymph nodes, lung, kidney and adrenal glands are examined histopathologically for tumor metastasis and the prostate tumor grade is also assessed.
  • Fiore C (2005b) Glycyrrhetinic acid, the active principle of licorice, can reduce the thickness of subcutaneous thigh fat through topical application. Steroids 70:538-542.
  • a synthetic triterpenoid, 2-cyano-3,12-dioxooleana-1 ,9-dien-28- oic acid (CDDO), is a ligand for the peroxisome proliferator-activated receptor g. MoI Endocrinol 14:1550-1556.
  • troglitazone Activation of peroxisome proliferator-activated receptor gamma by troglitazone inhibits cell growth through the increase of p27 KiP1 in human pancreatic carcinoma cells. Cancer Res. 60:5558-5564, 2000.
  • PPARg ligand induces growth arrest and differentiation markers of human pancreatic cancer cells. Int. J. Oncol.
  • DIM 3,3'-Diindolylmethane
  • derivatives induce apoptosis in pancreatic cancer cells through endoplasmic reticulum stress- dependent upregulation of DR5.
  • CDDO-Im 2-Cyano-3,12 dioxooleana-1 ,9 diene-28-imidazolide
  • Transcription factor Sp1 is essential for early embryonic development but dispensable for cell growth and differentiation. Cell 89:619-628, 1997.
  • VEGFR1 vascular endothelial growth factor receptor-1
  • Celecoxib inhibits vascular endothelial growth factor expression in and reduces angiogenesis and metastasis of human pancreatic cancer via suppression of Sp 1 transcription factor activity. Cancer Res. 64:2030- 2038, 2004.
  • NAG-1 a transforming growth factor-b superfamily member, by troglitazone requires the early growth response gene EGR-1. J Biol Chem 2004;279:6883-92.
  • Cyclooxygenase inhibitors induce the expression of the tumor suppressor gene EGR-1, which results in the up-regulation of NAG-1, an antitumorigenic protein.
  • EGR-1 tumor suppressor gene
  • 101. Ciccarelli C, Marampon F, Scoglio A, et al. p21 WAF1 expression induced by MEK/ERK pathway activation or inhibition correlates with growth arrest, myogenic differentiation and onco-phenotype reversal in rhabdomyosarcoma cells. MoI Cancer 2005;4:41.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Epidemiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Psychology (AREA)
  • Psychiatry (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Steroid Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

La présente invention concerne de nouveaux dérivés de l'acide glycyrrhétinique de formule (I), des compositions comprenant lesdits dérivés et l'utilisation de ceux-ci dans le traitement d'affections ou maladies, telles que le cancer, le diabète et la maladie de Huntington, qui bénéficient d'une régulation à la hausse du PPARγ et/ou d'une régulation à la baisse de l'expression ou de l'activité d'une ou plusieurs protéines de spécificité (Sp).
PCT/CA2007/001127 2006-06-27 2007-06-27 Dérivés de l'acide glycyrrhétinique WO2008000070A1 (fr)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US12/308,798 US8389573B2 (en) 2006-06-27 2007-06-27 Glycyrrhetinic acid derivatives
EP07720042.6A EP2032594B1 (fr) 2006-06-27 2007-06-27 Dérivés de l'acide glycyrrhétinique
JP2009516837A JP5610766B2 (ja) 2006-06-27 2007-06-27 グリチルレチン酸誘導体
CA2653726A CA2653726C (fr) 2006-06-27 2007-06-27 Derives de l'acide glycyrrhetinique
HK09107679.4A HK1128477A1 (zh) 2006-06-27 2009-08-20 甘草次酸衍生物
US13/759,320 US20130210926A1 (en) 2006-06-27 2013-02-05 Glycyrrhetinic acid derivatives

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US81659006P 2006-06-27 2006-06-27
US60/816,590 2006-06-27

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/759,320 Division US20130210926A1 (en) 2006-06-27 2013-02-05 Glycyrrhetinic acid derivatives

Publications (2)

Publication Number Publication Date
WO2008000070A1 true WO2008000070A1 (fr) 2008-01-03
WO2008000070B1 WO2008000070B1 (fr) 2008-03-27

Family

ID=38845067

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CA2007/001121 WO2008000068A1 (fr) 2006-06-27 2007-06-27 Procédé de préparation de composés 2-iodo triterpénoïdes
PCT/CA2007/001127 WO2008000070A1 (fr) 2006-06-27 2007-06-27 Dérivés de l'acide glycyrrhétinique

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/CA2007/001121 WO2008000068A1 (fr) 2006-06-27 2007-06-27 Procédé de préparation de composés 2-iodo triterpénoïdes

Country Status (6)

Country Link
US (2) US8389573B2 (fr)
EP (1) EP2032594B1 (fr)
JP (1) JP5610766B2 (fr)
CA (1) CA2653726C (fr)
HK (1) HK1128477A1 (fr)
WO (2) WO2008000068A1 (fr)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102241726A (zh) * 2011-05-27 2011-11-16 苏州大学 甘草次酸衍生物及其作为抗肿瘤药物的应用
CN102250189A (zh) * 2011-05-20 2011-11-23 中国药科大学 一种具有1,12-二烯-3-酮骨架的甘草次酸衍生物、其制备方法及医药用途
US8124656B2 (en) 2008-04-18 2012-02-28 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with saturation in the C-ring
US8124799B2 (en) 2008-04-18 2012-02-28 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with amino and other modifications at C-17
US8258329B2 (en) 2008-04-18 2012-09-04 Reata Pharmaceuticals, Inc. Dehydroandrosterone analogs including an anti-inflammatory pharmacore and methods of use
US8314137B2 (en) 2008-07-22 2012-11-20 Trustess Of Dartmouth College Monocyclic cyanoenones and methods of use thereof
US8394967B2 (en) 2008-04-18 2013-03-12 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: C-17 homologated oleanolic acid derivatives
EP2646045A1 (fr) * 2010-12-02 2013-10-09 Massachusetts Institute Of Technology Suppresseurs chimiques et de type arni de la neurotoxicité dans la maladie de huntington
US9278912B2 (en) 2012-09-10 2016-03-08 Reata Pharmaceuticals, Inc. C13-hydroxy derivatives of oleanolic acid and methods of use thereof
US9290536B2 (en) 2011-03-11 2016-03-22 Reata Pharmaceuticals, Inc. C4 monomethyl triterpenoid derivatives and methods of use thereof
US9512094B2 (en) 2012-09-10 2016-12-06 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US9701709B2 (en) 2012-04-27 2017-07-11 Reata Pharmaceuticals, Inc. 2,2-difluoropropionamide derivatives of bardoxolone methyl, polymorphic forms and methods of use thereof
US9757359B2 (en) 2008-01-11 2017-09-12 Reata Pharmaceuticals, Inc. Synthetic triterpenoids and methods of use in the treatment of disease
US9884809B2 (en) 2010-12-17 2018-02-06 Reata Pharmaceuticals, Inc. Pyrazolyl and pyrimidinyl tricyclic enones as antioxidant inflammation modulators
US10105372B2 (en) 2010-04-12 2018-10-23 Reata Pharmaceuticals, Inc. Methods of treating obesity using antioxidant inflammation modulators
CN110551169A (zh) * 2019-09-10 2019-12-10 陈昱西 甘草次酸类衍生物及其制备方法和用途
US10501489B2 (en) 2012-09-10 2019-12-10 Reata Pharmaceuticals, Inc. C17-alkanediyl and alkenediyl derivatives of oleanolic acid and methods of use thereof
US11584775B2 (en) 2015-09-23 2023-02-21 Reata Pharmaceuticals, Inc. C4-modified oleanolic acid derivatives for inhibition of IL-17 and other uses
US12060340B2 (en) 2018-06-20 2024-08-13 Reata Pharmaceuticals, Inc Cysteine-dependent inverse agonists of nuclear receptors ROR-gamma/ROR-gamma-t and methods of treating diseases or disorders therewith

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2032594B1 (fr) 2006-06-27 2015-09-23 Wellington Laboratories Inc. Dérivés de l'acide glycyrrhétinique
WO2013134558A1 (fr) 2012-03-07 2013-09-12 The Texas A & M University System Traitement du cancer ciblant la surexpression d'arn non codant
US8981144B2 (en) * 2012-05-08 2015-03-17 Trustees Of Dartmouth College Method for synthesizing 2-cyano-3,12-dioxoolean-1, 9(11)-dien-28-oic acid methyl ester and derivatives thereof
WO2013188818A1 (fr) 2012-06-15 2013-12-19 Reata Pharmaceuticals, Inc. Modulateurs anti-inflammation basés sur des triterpénoïdes époxydés au niveau du cycle a et leurs méthodes d'utilisation
US20160009755A1 (en) * 2013-03-01 2016-01-14 Revlon Consumer Products Corporation Cyrrhetinic alkyl esters and protected derivatives thereof
RU2611362C1 (ru) * 2015-11-26 2017-02-21 Федеральное государственное бюджетное учреждение науки Институт химической биологии и фундаментальной медицины Сибирского отделения Российской академии наук (ИХБФМ СО РАН) Водорастворимая композиция, обладающая противоопухолевой активностью и способ ее получения
CN106383188B (zh) * 2016-08-31 2018-08-28 岛津企业管理(中国)有限公司 一种齐墩果酸和熊果酸的定量检测方法
CN106543261A (zh) * 2016-10-27 2017-03-29 深圳市新阳唯康科技有限公司 一种甘草次酸晶型物质及其制备方法
CN106632575A (zh) * 2016-12-20 2017-05-10 深圳市新阳唯康科技有限公司 一种甘草次酸新晶型及其制备方法
WO2019024920A1 (fr) * 2017-08-03 2019-02-07 Medytox Inc. Procédés de préparation d'acides biliaires
JP2022519714A (ja) * 2019-02-07 2022-03-24 アルデリックス, インコーポレイテッド 高カリウム血症の治療で使用するためのグリチルレチン酸誘導体
CA3146842A1 (fr) * 2019-07-12 2021-01-21 Research Development Foundation Vaccins anti-ehrlichia et compositions immunogenes
CN114129572B (zh) * 2021-12-16 2023-04-07 南开大学 一种协同抑制汉防己甲素诱导的药源性肝损伤的药物组合物
CN115448973A (zh) * 2022-09-05 2022-12-09 承德医学院 具有柔性桥键结构的甘草次酸-叠氮类化合物、其制备方法和用途

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB887011A (en) * 1959-07-24 1962-01-10 Biorex Laboratories Ltd Glycyrrhetinic acid derivatives
GB1024802A (en) * 1963-05-30 1966-04-06 Biorex Laboratories Ltd New derivatives of glycyrrhetinic acid

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ191586A (en) 1978-10-10 1981-10-19 Sterling Drug Inc Cyanoketones derived from glycyrrhetinic acid and pharmaceutical compositions
EP2032594B1 (fr) 2006-06-27 2015-09-23 Wellington Laboratories Inc. Dérivés de l'acide glycyrrhétinique

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB887011A (en) * 1959-07-24 1962-01-10 Biorex Laboratories Ltd Glycyrrhetinic acid derivatives
GB1024802A (en) * 1963-05-30 1966-04-06 Biorex Laboratories Ltd New derivatives of glycyrrhetinic acid

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 2003
"The United States Pharmacopeia", 1999, THE NATIONAL FORMULARY
CHINTHARLAPALLI, S. ET AL., MOL. CANCER THERAP., vol. 6, 2007, pages 1588
ELGAMAL ET AL.: "Glycyrrhetic acid derivatives with modified ring A", J. PHARM. SCI., vol. 62, no. 9, 1973, pages 1557 - 1558, XP008102515 *
K. R. MORRIS: "Polymorphism in Pharmaceutical Solids", 1995, MARCEL DEKKER
KIM ET AL.: "Synthesis of new A-nor-derivatives of glycyrrhetic acid", ZHURNAL OBSHCHEI KHIMII, vol. 48, no. 9, 1978, pages 2146 - 2147, XP008103409 *
PITZELE: "Synthesis of 2-oxygenated glycyrrhetic acid derivatives", J. MED. CHEM., vol. 17, no. 2, 1974, pages 191 - 194, XP008102467 *
QIU, Z. ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 16672
RIBO ET AL.: "Synthesis of 1,11-dioxo-olean-2,12-dien-30-oic acid methyl ester and its 24-nor derivative", AFINIDAD, vol. 38, no. 373, 1981, pages 197 - 200, XP008100652 *
See also references of EP2032594A4

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9757359B2 (en) 2008-01-11 2017-09-12 Reata Pharmaceuticals, Inc. Synthetic triterpenoids and methods of use in the treatment of disease
US8440820B2 (en) 2008-04-18 2013-05-14 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with saturation in the C-ring
US10093614B2 (en) 2008-04-18 2018-10-09 Reata Pharmaceuticals, Inc. Antioxidant Inflamation modulators: oleanolic acid derivatives with amino and other modifications at C-17
US9796668B2 (en) 2008-04-18 2017-10-24 Reata Pharmaceuticals, Inc. Natural product analogs including an anti-inflammatory cyanoenone pharmacore and methods of use
US8258329B2 (en) 2008-04-18 2012-09-04 Reata Pharmaceuticals, Inc. Dehydroandrosterone analogs including an anti-inflammatory pharmacore and methods of use
US11091430B2 (en) 2008-04-18 2021-08-17 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with amino and other modifications at c-17
US8394967B2 (en) 2008-04-18 2013-03-12 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: C-17 homologated oleanolic acid derivatives
US8440854B2 (en) 2008-04-18 2013-05-14 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with amino acid and other modifications at C-17
US9670147B2 (en) 2008-04-18 2017-06-06 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with amino and other modifications at C-17
US8124799B2 (en) 2008-04-18 2012-02-28 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with amino and other modifications at C-17
US8124656B2 (en) 2008-04-18 2012-02-28 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with saturation in the C-ring
US11919838B2 (en) 2008-04-18 2024-03-05 Reata Pharmaceuticals Holdings, LLC Antioxidant inflammation modulators: oleanolic acid derivatives with amino and other modifications at C-17
US9090574B2 (en) 2008-04-18 2015-07-28 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with saturation in the C-ring
US10556858B2 (en) 2008-04-18 2020-02-11 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with amino and other modifications at C-17
US8314137B2 (en) 2008-07-22 2012-11-20 Trustess Of Dartmouth College Monocyclic cyanoenones and methods of use thereof
US9000188B2 (en) 2008-07-22 2015-04-07 Trustees Of Dartmouth College Monocyclic cyanoenones and methods of use thereof
US10105372B2 (en) 2010-04-12 2018-10-23 Reata Pharmaceuticals, Inc. Methods of treating obesity using antioxidant inflammation modulators
US11911395B2 (en) 2010-04-12 2024-02-27 Reata Pharmaceuticals Holdings, LLC Methods of treating obesity using antioxidant inflammation modulators
EP2646045A4 (fr) * 2010-12-02 2014-05-07 Massachusetts Inst Technology Suppresseurs chimiques et de type arni de la neurotoxicité dans la maladie de huntington
EP2646045A1 (fr) * 2010-12-02 2013-10-09 Massachusetts Institute Of Technology Suppresseurs chimiques et de type arni de la neurotoxicité dans la maladie de huntington
US9884809B2 (en) 2010-12-17 2018-02-06 Reata Pharmaceuticals, Inc. Pyrazolyl and pyrimidinyl tricyclic enones as antioxidant inflammation modulators
US11814338B2 (en) 2010-12-17 2023-11-14 Reata Pharmaceuticals, Inc. Pyrazolyl and pyrimidinyl tricyclic enones as antioxidant inflammation modulators
US11192852B2 (en) 2010-12-17 2021-12-07 Reata Pharmaceuticals, Inc. Pyrazolyl and pyrimidinyl tricyclic enones as antioxidant inflammation modulators
US9290536B2 (en) 2011-03-11 2016-03-22 Reata Pharmaceuticals, Inc. C4 monomethyl triterpenoid derivatives and methods of use thereof
CN102250189A (zh) * 2011-05-20 2011-11-23 中国药科大学 一种具有1,12-二烯-3-酮骨架的甘草次酸衍生物、其制备方法及医药用途
CN102241726A (zh) * 2011-05-27 2011-11-16 苏州大学 甘草次酸衍生物及其作为抗肿瘤药物的应用
US11078230B2 (en) 2012-04-27 2021-08-03 Reata Pharmaceuticals, Inc. 2,2-difluoropropionamide derivatives of bardoxolone methyl, polymorphic forms and methods of use thereof
US12065464B2 (en) 2012-04-27 2024-08-20 Reata Pharmaceuticals Holdings, LLC 2,2-difluoropropionamide derivatives of bardoxolone methyl, polymorphic forms and methods of use thereof
US9701709B2 (en) 2012-04-27 2017-07-11 Reata Pharmaceuticals, Inc. 2,2-difluoropropionamide derivatives of bardoxolone methyl, polymorphic forms and methods of use thereof
US10398711B2 (en) 2012-09-10 2019-09-03 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US10898499B2 (en) 2012-09-10 2021-01-26 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US9278912B2 (en) 2012-09-10 2016-03-08 Reata Pharmaceuticals, Inc. C13-hydroxy derivatives of oleanolic acid and methods of use thereof
US11406648B2 (en) 2012-09-10 2022-08-09 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US10501489B2 (en) 2012-09-10 2019-12-10 Reata Pharmaceuticals, Inc. C17-alkanediyl and alkenediyl derivatives of oleanolic acid and methods of use thereof
US9512094B2 (en) 2012-09-10 2016-12-06 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US9889143B2 (en) 2012-09-10 2018-02-13 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US11584775B2 (en) 2015-09-23 2023-02-21 Reata Pharmaceuticals, Inc. C4-modified oleanolic acid derivatives for inhibition of IL-17 and other uses
US12060340B2 (en) 2018-06-20 2024-08-13 Reata Pharmaceuticals, Inc Cysteine-dependent inverse agonists of nuclear receptors ROR-gamma/ROR-gamma-t and methods of treating diseases or disorders therewith
CN110551169B (zh) * 2019-09-10 2022-07-15 陈昱西 甘草次酸类衍生物及其制备方法和用途
CN110551169A (zh) * 2019-09-10 2019-12-10 陈昱西 甘草次酸类衍生物及其制备方法和用途

Also Published As

Publication number Publication date
EP2032594A4 (fr) 2012-09-05
JP2009541364A (ja) 2009-11-26
EP2032594B1 (fr) 2015-09-23
HK1128477A1 (zh) 2009-10-30
US20130210926A1 (en) 2013-08-15
CA2653726A1 (fr) 2008-01-03
WO2008000070B1 (fr) 2008-03-27
EP2032594A1 (fr) 2009-03-11
US8389573B2 (en) 2013-03-05
WO2008000068A1 (fr) 2008-01-03
CA2653726C (fr) 2015-05-12
JP5610766B2 (ja) 2014-10-22
US20100099760A1 (en) 2010-04-22

Similar Documents

Publication Publication Date Title
EP2032594B1 (fr) Dérivés de l'acide glycyrrhétinique
H Safe et al. Unifying mechanisms of action of the anticancer activities of triterpenoids and synthetic analogs
CA2912139C (fr) Derives 11-hydroxyle d'acides biliaires et leurs conjugues d'acides amines en tant que modulateurs du recepteur de farnesoide x
KR101739800B1 (ko) 안드로겐 수용체 조절제의 에스터 유도체 및 그의 이용 방법
Chintharlapalli et al. Structure-dependent activity of glycyrrhetinic acid derivatives as peroxisome proliferator–activated receptor γ agonists in colon cancer cells
Mansure et al. Peroxisome proliferator-activated receptor γ in bladder cancer: a promising therapeutic target
US20090203661A1 (en) Betulinic acid, derivatives and analogs thereof and uses therefor
Shi et al. Novel anti-prostate cancer curcumin analogues that enhance androgen receptor degradation activity
CN107001406B (zh) 用于在肿瘤的治疗中使用的11-脱氧皮质醇的17α,21-二酯
CA2372493A1 (fr) Derives steroides
Ju et al. Design and synthesis of novel mitochondria-targeted CDDO derivatives as potential anti-cancer agents
US10487056B2 (en) Small molecule androgen receptor inhibitors and methods of use thereof
Al-Kazaale et al. Synthesis, molecular modeling and biological evaluation of potent analogs of 2-methoxyestradiol
López-Huerta et al. Synthesis, biological evaluation, molecular docking studies and In-silico ADMET evaluation of pyrazines of pentacyclic triterpenes
US20150148321A1 (en) Substituted androst-4-ene diones
US8575141B2 (en) Steroid-derived cyclopamine analogs and methods for using the same in the prevention or treatment of cancer
WO2018175965A1 (fr) Anti-œstrogènes de thérapie du cancer du sein
JP5955531B2 (ja) 抗癌剤
US8669243B2 (en) Steroid-derived cyclopamine analogs and methods for using the same in the prevention or treatment of cancer
NZ714082B2 (en) 11-hydroxyl-derivatives of bile acids and amino acid conjugates thereof as farnesoid x receptor modulators

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07720042

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2653726

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2009516837

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007720042

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: RU

WWE Wipo information: entry into national phase

Ref document number: 12308798

Country of ref document: US